Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Nanotoxicology ; 18(1): 69-86, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38420937

ABSTRACT

In the lung, carcinogenesis is a multi-stage process that includes initiation by a genotoxic agent, promotion that expands the population of cells with damaged DNA to form a tumor, and progression from benign to malignant neoplasms. We have previously shown that Mitsui-7, a long and rigid multi-walled carbon nanotube (MWCNT), promotes pulmonary carcinogenesis in a mouse model. To investigate the potential exposure threshold and dose-response for tumor promotion by this MWCNT, 3-methylcholanthrene (MC) initiated (10 µg/g, i.p., once) or vehicle (corn oil) treated B6C3F1 mice were exposed by inhalation to filtered air or MWCNT (5 mg/m3) for 5 h/day for 0, 2, 5, or 10 days and were followed for 17 months post-exposure for evidence of lung tumors. Pulmonary neoplasia incidence in MC-initiated mice significantly increased with each MWCNT exposure duration. Exposure to either MC or MWCNT alone did not affect pulmonary neoplasia incidence compared with vehicle controls. Lung tumor multiplicity in MC-initiated mice also significantly increased with each MWCNT exposure duration. Thus, a significantly higher lung tumor multiplicity was observed after a 10-day MWCNT exposure than following a 2-day exposure. Both bronchioloalveolar adenoma and bronchioloalveolar adenocarcinoma multiplicity in MC-initiated mice were significantly increased following 5- and 10-day MWCNT exposure, while a 2-day MWCNT exposure in MC-initiated mice significantly increased the multiplicity of adenomas but not adenocarcinomas. In this study, even the lowest MWCNT exposure promoted lung tumors in MC-initiated mice. Our findings indicate that exposure to this MWCNT strongly promotes pulmonary carcinogenesis.


Subject(s)
Lung Neoplasms , Lung , Mice , Animals , Lung/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Mice, Inbred Strains , Cell Transformation, Neoplastic , Carcinogenesis/chemically induced , Carcinogenesis/pathology , Inhalation Exposure , Mice, Inbred C57BL
2.
Small ; 18(52): e2203259, 2022 12.
Article in English | MEDLINE | ID: mdl-36373669

ABSTRACT

The toxicity of boron nitride nanotubes (BNNTs) has been the subject of conflicting reports, likely due to differences in the residuals and impurities that can make up to 30-60% of the material produced based on the manufacturing processes and purification employed. Four BNNTs manufactured by induction thermal plasma process with a gradient of BNNT purity levels achieved through sequential gas purification, water and solvent washing, allowed assessing the influence of these residuals/impurities on the toxicity profile of BNNTs. Extensive characterization including infrared and X-ray spectroscopy, thermogravimetric analysis, size, charge, surface area, and density captured the alteration in physicochemical properties as the material went through sequential purification. The material from each step is screened using acellular and in vitro assays for evaluating general toxicity, mechanisms of toxicity, and macrophage function. As the material increased in purity, there are more high-aspect-ratio particulates and a corresponding distinct increase in cytotoxicity, nuclear factor-κB transcription, and inflammasome activation. There is no alteration in macrophage function after BNNT exposure with all purity grades. The cytotoxicity and mechanism of screening clustered with the purity grade of BNNTs, illustrating that greater purity of BNNT corresponds to greater toxicity.


Subject(s)
Boron Compounds , Nanotubes , Boron Compounds/toxicity , Boron Compounds/chemistry , Macrophages , Nanotubes/toxicity , Nanotubes/chemistry
3.
Front Immunol ; 11: 1186, 2020.
Article in English | MEDLINE | ID: mdl-32595644

ABSTRACT

Pulmonary exposure to certain engineered nanomaterials (ENMs) causes chronic lesions like fibrosis and cancer in animal models as a result of unresolved inflammation. Resolution of inflammation involves the time-dependent biosynthesis of lipid mediators (LMs)-in particular, specialized pro-resolving mediators (SPMs). To understand how ENM-induced pulmonary inflammation is resolved, we analyzed the inflammatory and pro-resolving responses to fibrogenic multi-walled carbon nanotubes (MWCNTs, Mitsui-7) and low-toxicity fullerenes (fullerene C60, C60F). Pharyngeal aspiration of MWCNTs at 40 µg/mouse or C60F at a dose above 640 µg/mouse elicited pulmonary effects in B6C3F1 mice. Both ENMs stimulated acute inflammation, predominated by neutrophils, in the lung at day 1, which transitioned to histiocytic inflammation by day 7. By day 28, the lesion in MWCNT-exposed mice progressed to fibrotic granulomas, whereas it remained as alveolar histiocytosis in C60F-exposed mice. Flow cytometric profiling of whole lung lavage (WLL) cells revealed that neutrophil recruitment was the greatest at day 1 and declined to 36.6% of that level in MWCNT- and 16.8% in C60F-treated mice by day 7, and to basal levels by day 28, suggesting a rapid initiation phase and an extended resolution phase. Both ENMs induced high levels of proinflammatory leukotriene B4 (LTB4) and prostaglandin E2 (PGE2) with peaks at day 1, and high levels of SPMs resolvin D1 (RvD1) and E1 (RvE1) with peaks at day 7. MWCNTs and C60F induced time-dependent polarization of M1 macrophages with a peak at day 1 and subsequently of M2 macrophages with a peak at day 7 in the lung, accompanied by elevated levels of type 1 or type 2 cytokines, respectively. M1 macrophages exhibited preferential induction of arachidonate 5-lipoxygenase activating protein (ALOX5AP), whereas M2 macrophages had a high level expression of arachidonate 15-lipoxygenase (ALOX15). Polarization of macrophages in vitro differentially induced ALOX5AP in M1 macrophages or ALOX15 in M2 macrophages resulting in increased preferential biosynthesis of proinflammatory LMs or SPMs. MWCNTs increased the M1- or M2-specific production of LMs accordingly. These findings support a mechanism by which persistent ENM-induced neutrophilic inflammation is actively resolved through time-dependent polarization of macrophages and enhanced biosynthesis of specialized LMs via distinct ALOX pathways.


Subject(s)
Fullerenes/toxicity , Macrophage Activation/immunology , Macrophages/immunology , Nanotubes, Carbon/toxicity , Pneumonia/chemically induced , Pneumonia/immunology , Animals , Macrophages/drug effects , Mice , Pneumonia/pathology
4.
Inhal Toxicol ; 32(1): 24-38, 2020 01.
Article in English | MEDLINE | ID: mdl-32028803

ABSTRACT

Objective: In this study, we compared in vitro and in vivo bioactivity of nitrogen-doped multi-walled carbon nanotubes (NDMWCNT) to MWCNT to test the hypothesis that nitrogen doping would alter bioactivity.Materials and Methods: High-resolution transmission electron microscopy (TEM) confirmed the multilayer structure of MWCNT with an average layer distance of 0.36 nm, which was not altered by nitrogen doping: the nanomaterials had similar widths and lengths. In vitro studies with THP-1 cells and alveolar macrophages from C57BL/6 mice demonstrated that NDMWCNT were less cytotoxic and stimulated less IL-1ß release compared to MWCNT. For in vivo studies, male C57BL/6J mice received a single dose of dispersion medium (DM), 2.5, 10 or 40 µg/mouse of NDMWCNT, or 40 µg/mouse of MWCNT by oropharyngeal aspiration. Animals were euthanized between 1 and 7 days post-exposure for whole lung lavage (WLL) studies.Results and Discussion: NDMWCNT caused time- and dose-dependent pulmonary inflammation. However, it was less than that caused by MWCNT. Activation of the NLRP3 inflammasome was assessed in particle-exposed mice by determining cytokine production in WLL fluid at 1 day post-exposure. Compared to DM-exposed mice, IL-1ß and IL-18 were significantly increased in MWCNT- and NDMWCNT-exposed mice, but the increase caused by NDMWCNT was less than MWCNT. At 56 days post-exposure, histopathology determined lung fibrosis in MWCNT-exposed mice was greater than NDMWCNT-exposed mice.Conclusions: These data indicate nitrogen doping of MWCNT decreases their bioactivity, as reflected with lower in vitro and in vivo toxicity inflammation and lung disease. The lower activation of the NLRP3 inflammasome may be responsible. Abbreviations: NDMWCNT: nitrogen-doped multi-walled carbon nanotubes; MWCNT: multi-walled carbon nanotubes; TEM: transmission electron microscopy; HRTEM: high resolution transmission electron microscopy; IL-1ß: interleukin-1ß; DM: dispersion medium; WLL: whole lung lavage; IL-18: interleukin-18; GSD: geometric standard deviation; XPS: X-ray photoelectron spectroscopy; SEM: standard error of the mean; PMA: phorbol 12-myristate 13-acetate; LPS: lipopolysacharride; LDH: lactate dehydrogenase; AM: alveolar macrophage; PMN: polymorphonuclear leukocyte.


Subject(s)
Inhalation Exposure/adverse effects , Lung/drug effects , Macrophages, Alveolar/drug effects , Nanotubes, Carbon/toxicity , Nitrogen/toxicity , Pneumonia/chemically induced , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cytokines/analysis , Dose-Response Relationship, Drug , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Lung/immunology , Lung/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Male , Mice, Inbred C57BL , Nanotubes, Carbon/chemistry , Nitrogen/chemistry , Particle Size , Pneumonia/immunology , Pneumonia/pathology , Surface Properties , THP-1 Cells , Time Factors
5.
Int J Mol Sci ; 20(11)2019 May 29.
Article in English | MEDLINE | ID: mdl-31146342

ABSTRACT

As the demand for multi-walled carbon nanotube (MWCNT) incorporation into industrial and biomedical applications increases, so does the potential for unintentional pulmonary MWCNT exposure, particularly among workers during manufacturing. Pulmonary exposure to MWCNTs raises the potential for development of lung inflammation, fibrosis, and cancer among those exposed; however, there are currently no effective biomarkers for detecting lung fibrosis or predicting the risk of lung cancer resulting from MWCNT exposure. To uncover potential mRNAs and miRNAs that could be used as markers of exposure, this study compared in vivo mRNA and miRNA expression in lung tissue and blood of mice exposed to MWCNTs with in vitro mRNA and miRNA expression from a co-culture model of human lung epithelial and microvascular cells, a system previously shown to have a higher overall genome-scale correlation with mRNA expression in mouse lungs than either cell type grown separately. Concordant mRNAs and miRNAs identified by this study could be used to drive future studies confirming human biomarkers of MWCNT exposure. These potential biomarkers could be used to assess overall worker health and predict the occurrence of MWCNT-induced diseases.


Subject(s)
Lung Diseases/blood , Lung/metabolism , MicroRNAs/blood , Nanotubes, Carbon/toxicity , RNA, Messenger/blood , Animals , Biomarkers/blood , Biomarkers/metabolism , Cell Line , Cells, Cultured , Humans , Lung/drug effects , Lung Diseases/etiology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Occupational Exposure , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Int J Toxicol ; 37(4): 276-284, 2018.
Article in English | MEDLINE | ID: mdl-29916280

ABSTRACT

Respiratory exposure to multiwalled carbon nanotubes (MWCNT) or asbestos results in fibrosis; however, the mechanisms to reach this end point may be different. A previous study by our group identified pulmonary effects and significantly altered messenger RNA (mRNA) signaling pathways following exposure to 1, 10, 40, and 80 µg MWCNT and 120 µg crocidolite asbestos on mouse lungs over time at 1-month, 6-month, and 1-year postexposure following pulmonary aspiration. As a continuation to the above study, this current study took an in-depth look at the signaling pathways involved in fibrosis development at a single time point, 1 year, and exposure, 40 µg MWCNT, the lowest exposure at which fibrosis was pathologically evident. The 120 µg asbestos exposure was included to compare MWCNT-induced fibrosis with asbestos-induced fibrosis. A previously validated computational model was used to identify mRNAs with expression profiles matching the fibrosis pathology patterns from exposed mouse lungs. mRNAs that matched the pathology patterns were then input into ingenuity pathway analysis to determine potential signaling pathways and physiological disease functions inherent to MWCNT and asbestos exposure. Both MWCNT and asbestos exposure induced changes in mouse lungs regarding gene expression, cell proliferation, and survival, while MWCNT uniquely induced alterations in pathways involved in oxidative phosphorylation, mitochondrial dysfunction, and transcription. Asbestos exposure produced unique alterations in pathways involved in sustained inflammation. Although typically considered similar due to scale and fiber-like appearance, the different compositional properties inherent to either MWCNT or asbestos may play a role in their ability to induce fibrosis after pulmonary exposure.


Subject(s)
Asbestos, Crocidolite/toxicity , Nanotubes, Carbon/toxicity , Pulmonary Fibrosis/chemically induced , Administration, Inhalation , Animals , Gene Expression/drug effects , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , RNA, Messenger/metabolism
7.
Nanotoxicology ; 11(8): 1040-1058, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29094619

ABSTRACT

Boron nitride nanotubes (BNNTs) are an emerging engineered nanomaterial attracting significant attention due to superior electrical, chemical and thermal properties. Currently, the toxicity profile of this material is largely unknown. Commercial grade BNNTs are composed of a mixture (BNNT-M) of ∼50-60% BNNTs, and ∼40-50% impurities of boron and hexagonal boron nitride. We performed acute in vitro and in vivo studies with commercial grade BNNT-M, dispersed by sonication in vehicle, in comparison to the extensively studied multiwalled carbon nanotube-7 (MWCNT-7). THP-1 wild-type and NLRP3-deficient human monocytic cells were exposed to 0-100 µg/ml and C57BL/6 J male mice were treated with 40 µg of BNNT-M for in vitro and in vivo studies, respectively. In vitro, BNNT-M induced a dose-dependent increase in cytotoxicity and oxidative stress. This was confirmed in vivo following acute exposure increase in bronchoalveolar lavage levels of lactate dehydrogenase, pulmonary polymorphonuclear cell influx, loss in mitochondrial membrane potential and augmented levels of 4-hydroxynonenal. Uptake of this material caused lysosomal destabilization, pyroptosis and inflammasome activation, corroborated by an increase in cathepsin B, caspase 1, increased protein levels of IL-1ß and IL-18 both in vitro and in vivo. Attenuation of these effects in NLRP3-deficient THP-1 cells confirmed NLRP3-dependent inflammasome activation by BNNT-M. BNNT-M induced a similar profile of inflammatory pulmonary protein production when compared to MWCNT-7. Functionally, pretreatment with BNNT-M caused suppression in bacterial uptake by THP-1 cells, an effect that was mirrored in challenged alveolar macrophages collected from exposed mice and attenuated with NLRP3 deficiency. Analysis of cytokines secreted by LPS-challenged alveolar macrophages collected after in vivo exposure to dispersions of BNNT-M showed a differential macrophage response. The observed results demonstrated acute inflammation and toxicity in vitro and in vivo following exposure to sonicated BNNT-M was in part due to NLRP3 inflammasome activation.


Subject(s)
Boron Compounds/toxicity , Lung/drug effects , Nanotubes/toxicity , Oxidative Stress/drug effects , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Inflammation , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Particle Size , Pyroptosis/drug effects
8.
Part Fibre Toxicol ; 13(1): 34, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27328692

ABSTRACT

BACKGROUND: Graphene, a monolayer of carbon, is an engineered nanomaterial (ENM) with physical and chemical properties that may offer application advantages over other carbonaceous ENMs, such as carbon nanotubes (CNT). The goal of this study was to comparatively assess pulmonary and systemic toxicity of graphite nanoplates, a member of the graphene-based nanomaterial family, with respect to nanoplate size. METHODS: Three sizes of graphite nanoplates [20 µm lateral (Gr20), 5 µm lateral (Gr5), and <2 µm lateral (Gr1)] ranging from 8-25 nm in thickness were characterized for difference in surface area, structure,, zeta potential, and agglomeration in dispersion medium, the vehicle for in vivo studies. Mice were exposed by pharyngeal aspiration to these 3 sizes of graphite nanoplates at doses of 4 or 40 µg/mouse, or to carbon black (CB) as a carbonaceous control material. At 4 h, 1 day, 7 days, 1 month, and 2 months post-exposure, bronchoalveolar lavage was performed to collect fluid and cells for analysis of lung injury and inflammation. Particle clearance, histopathology and gene expression in lung tissue were evaluated. In addition, protein levels and gene expression were measured in blood, heart, aorta and liver to assess systemic responses. RESULTS: All Gr samples were found to be similarly composed of two graphite structures and agglomerated to varying degrees in DM in proportion to the lateral dimension. Surface area for Gr1 was approximately 7-fold greater than Gr5 and Gr20, but was less reactive reactive per m(2). At the low dose, none of the Gr materials induced toxicity. At the high dose, Gr20 and Gr5 exposure increased indices of lung inflammation and injury in lavage fluid and tissue gene expression to a greater degree and duration than Gr1 and CB. Gr5 and Gr20 showed no or minimal lung epithelial hypertrophy and hyperplasia, and no development of fibrosis by 2 months post-exposure. In addition, the aorta and liver inflammatory and acute phase genes were transiently elevated in Gr5 and Gr20, relative to Gr1. CONCLUSIONS: Pulmonary and systemic toxicity of graphite nanoplates may be dependent on lateral size and/or surface reactivity, with the graphite nanoplates > 5 µm laterally inducing greater toxicity which peaked at the early time points post-exposure relative to the 1-2 µm graphite nanoplate.


Subject(s)
Graphite/toxicity , Lung/drug effects , Nanoparticles , Nanostructures/toxicity , Animals , Bronchoalveolar Lavage Fluid , Lung/metabolism , Mice , Microscopy, Electron, Scanning , RNA, Messenger/metabolism
9.
J Toxicol Environ Health A ; 79(8): 352-66, 2016.
Article in English | MEDLINE | ID: mdl-27092743

ABSTRACT

Pulmonary exposure to multiwalled carbon nanotubes (MWCNT) induces an inflammatory and rapid fibrotic response, although the long-term signaling mechanisms are unknown. The aim of this study was to examine the effects of 1, 10, 40, or 80 µg MWCNT administered by pharyngeal aspiration on bronchoalveolar lavage (BAL) fluid for polymorphonuclear cell (PMN) infiltration, lactate dehydrogenase (LDH) activity, and lung histopathology for inflammatory and fibrotic responses in mouse lungs 1 mo, 6 mo, and 1 yr postexposure. Further, a 120-µg crocidolite asbestos group was incorporated as a positive control for comparative purposes. Results showed that MWCNT increased BAL fluid LDH activity and PMN infiltration in a dose-dependent manner at all three postexposure times. Asbestos exposure elevated LDH activity at all 3 postexposure times and PMN infiltration at 1 mo and 6 mo postexposure. Pathological changes in the lung, the presence of MWCNT or asbestos, and fibrosis were noted at 40 and 80 µg MWCNT and in asbestos-exposed mice at 1 yr postexposure. To determine potential signaling pathways involved with MWCNT-associated pathological changes in comparison to asbestos, up- and down-regulated gene expression was determined in lung tissue at 1 yr postexposure. Exposure to MWCNT tended to favor those pathways involved in immune responses, specifically T-cell responses, whereas exposure to asbestos tended to favor pathways involved in oxygen species production, electron transport, and cancer. Data indicate that MWCNT are biopersistent in the lung and induce inflammatory and fibrotic pathological alterations similar to those of crocidolite asbestos, but may reach these endpoints by different mechanisms.


Subject(s)
Air Pollutants/toxicity , Asbestos, Crocidolite/toxicity , Inhalation Exposure , Lung/drug effects , Lung/pathology , Nanotubes, Carbon/toxicity , Animals , Bronchoalveolar Lavage Fluid/chemistry , Dose-Response Relationship, Drug , Gene Expression/drug effects , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Time Factors
10.
Arch Toxicol ; 89(4): 621-33, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25510677

ABSTRACT

Multi-walled carbon nanotubes (MWCNT) are new materials with a wide range of industrial and commercial applications. However, their nano-scaled size and fiber-like shape render them respirable and potentially fibrogenic if inhaled into the lungs. To understand MWCNT fibrogenesis, we analyzed the pathologic and molecular aspects of the early phase response to MWCNT in mouse lungs. MWCNT induced rapid and pronounced lesions in the lungs characterized by increased cellularity and formation of fibrotic foci, most notably near where MWCNT deposited, within 14 days post-exposure. Deposition of collagen fibers was markedly increased in the alveolar septa and fibrotic foci, accompanied by elevated expression of fibrotic genes Col1a1, Col1a2, and Fn1 at both mRNA and protein levels. Fibrosis was induced rapidly at 40 µg, wherein fibrotic changes were detected on day 1 and reached a maximal intensity on day 7 through day 14. Induction of fibrosis was dose-dependent at the dose range of 5-40 µg, 7 days post-exposure. MWCNT elicited rapid and prominent infiltrations of neutrophils and macrophages alongside fibrosis implicating acute inflammation in the fibrotic response. At the molecular level, MWCNT induced elevated expression of proinflammatory cytokines TNFα, IL1α, IL1ß, IL6, and CCL2 in lung tissues as well as the bronchoalveolar lavage fluid, in a dose- and time-dependent manner. MWCNT also increased the expression of fibrogenic growth factors TGF-ß1 and PDGF-A in the lungs significantly. These findings underscore the interplay between acute inflammation and the early fibrotic response in the initiation and propagation of pulmonary fibrosis induced by MWCNT.


Subject(s)
Inhalation Exposure/adverse effects , Lung/drug effects , Nanotubes, Carbon/toxicity , Pulmonary Fibrosis , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Cytokines/analysis , Fibronectins/genetics , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Time Factors
11.
Toxicology ; 328: 66-74, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25511174

ABSTRACT

There is a current interest in reducing the in vivo toxicity testing of nanomaterials in animals by increasing toxicity testing using in vitro cellular assays; however, toxicological results are seldom concordant between in vivo and in vitro models. This study compared global multi-walled carbon nanotube (MWCNT)-induced gene expression from human lung epithelial and microvascular endothelial cells in monoculture and coculture with gene expression from mouse lungs exposed to MWCNT. Using a cutoff of 10% false discovery rate and 1.5 fold change, we determined that there were more concordant genes (gene expression both up- or downregulated in vivo and in vitro) expressed in both cell types in coculture than in monoculture. When reduced to only those genes involved in inflammation and fibrosis, known outcomes of in vivo MWCNT exposure, there were more disease-related concordant genes expressed in coculture than monoculture. Additionally, different cellular signaling pathways are activated in response to MWCNT dependent upon culturing conditions. As coculture gene expression better correlated with in vivo gene expression, we suggest that cellular cocultures may offer enhanced in vitro models for nanoparticle risk assessment and the reduction of in vivo toxicological testing.


Subject(s)
Endothelial Cells/drug effects , Epithelial Cells/drug effects , Lung/blood supply , Lung/drug effects , Nanotubes, Carbon/toxicity , Animals , Cells, Cultured , Coculture Techniques , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Genetic Markers , Humans , Inhalation Exposure/adverse effects , Lung/metabolism , Male , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Reproducibility of Results , Risk Assessment
12.
Toxicol Sci ; 144(1): 51-64, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25527334

ABSTRACT

Multi-walled carbon nanotubes (MWCNTs) are known for their transient inflammatory and progressive fibrotic pulmonary effects; however, the mechanisms underlying these pathologies are unknown. In this study, we used time-series microarray data of global lung mRNA and miRNA expression isolated from C57BL/6J mice exposed by pharyngeal aspiration to vehicle or 10, 20, 40, or 80 µg MWCNT at 1, 7, 28, or 56 days post-exposure to determine miRNA and mRNA regulatory networks that are potentially involved in MWCNT-induced inflammatory and fibrotic lung etiology. Using a non-negative matrix factorization method, we determined mRNAs and miRNAs with expression profiles associated with pathology patterns of MWCNT-induced inflammation (based on bronchoalveolar lavage score) and fibrosis (based on Sirius Red staining measured with quantitative morphometric analysis). Potential binding targets between pathology-related mRNAs and miRNAs were identified using Ingenuity Pathway Analysis and the miRTarBase, miRecords, and TargetScan databases. Using these experimentally validated and predicted binding targets, we were able to build molecular signaling networks that are potentially reflective of and play a role in MWCNT-induced lung inflammatory and fibrotic pathology. As understanding the regulatory networks between mRNAs and miRNAs in different disease states would be beneficial for understanding the complex mechanisms of pathogenesis, these identified genes and pathways may be useful for determining biomarkers of MWCNT-induced lung inflammation and fibrosis for early detection of disease.


Subject(s)
Gene Regulatory Networks , Genetic Markers , Lung/metabolism , MicroRNAs/genetics , Nanotubes, Carbon , Pneumonia/genetics , Pulmonary Fibrosis/genetics , RNA, Messenger/genetics , Animals , Computational Biology , Databases, Genetic , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Inflammation Mediators/metabolism , Inhalation Exposure , Lung/pathology , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , RNA, Messenger/metabolism , Time Factors
13.
Part Fibre Toxicol ; 10: 33, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23895460

ABSTRACT

BACKGROUND: Prior studies have demonstrated a rapid and progressive acute phase response to bolus aspiration of multi-walled carbon nanotubes (MWCNTs). In this study we sought to test the hypothesis that inhalation exposure to MWCNT produces a fibrotic response and that the response is chronically persistent. To address the hypothesis that inhaled MWCNTs cause persistent morphologic changes, male C57BL/6 J mice were exposed in a whole-body inhalation system to a MWCNT aerosol and the fibrotic response in the alveolar region examined at up to 336 days after termination of exposure. METHODS: Inhalation exposure was to a 5 mg/m3 MWCNT aerosol for 5 hours/day for 12 days (4 times/week for 3 weeks). At the end of inhalation exposures, lungs were either lavaged for analysis of bronchoalveolar lavage (BAL) or preserved by vascular perfusion of fixative while inflated with air at 1, 14, 84, 168 and 336 days post inhalation exposure. Separate, clean-air control groups were also studied. Light microscopy, enhanced darkfield microscopy and field emission electron microscopy (FESEM) of tissue sections were used to analyze the distribution of lung burden following inhalation exposure. Morphometric measurements of Sirius Red staining for fibrillar collagen were used to assess the connective tissue response. Serial section analysis of enhanced darkfield microscope images was used to examine the redistribution of MWCNT fibers within the lungs during the post-exposure period. RESULTS: At day 1 post-exposure 84 ± 3 and 16 ± 2 percent of the lung burden (Mean ± S.E., N = 5) were in the alveolar and airway regions, respectively. Initial distribution within the alveolar region was 56 ± 5, 7 ± 4 and 20 ± 3 percent of lung burden in alveolar macrophages, alveolar airspaces and alveolar tissue, respectively. Clearance reduced the alveolar macrophage burden of MWCNTs by 35 percent between 1 and 168 days post-exposure, while the content of MWCNTs in the alveolar tissue increased by 63 percent. Large MWCNT structures containing greater than 4 fibers were 53.6 percent of the initial lung burden and accounted for the majority of the decline with clearance, while lung burden of singlet MWCNT was essentially unchanged. The mean linear intercept of alveolar airspace, a measure of the expansion of the lungs, was not significantly different between groups. Pulmonary inflammation and damage, measured as the number of polymorphnuclear leukocytes (PMNs) or lactate dehydrogenase activity (LDH) and albumin in BAL, increased rapidly (1 day post) after inhalation of MWCNTs and declined slowly with time post-exposure. The fibrillar collagen in the alveolar region of MWCNT-exposed mice demonstrated a progressive increase in thickness over time (0.17 ± 0.02, 0.22 ± 0.02, 0.26 ± 0.03, 0.25 ± 0.02 and 0.29 ± 0.01 microns for 1, 14, 84, 168 and 336 days post-exposure) and was significantly different from clean-air controls (0.16 ± 0.02) at 84 and (0.15 ± 0.02) at 336 days post-exposure. CONCLUSIONS: Despite the relatively low fraction of the lung burden being delivered to the alveolar tissue, the average thickness of connective tissue in the alveolar region increased by 70% in the 336 days after inhalation exposure. These results demonstrate that inhaled MWCNTs deposit and are retained within the alveolar tissue where they produce a progressive and persistent fibrotic response up to 336 days post-exposure.


Subject(s)
Inhalation Exposure/adverse effects , Nanotubes, Carbon/toxicity , Pulmonary Alveoli/drug effects , Pulmonary Fibrosis/chemically induced , Aerosols , Albumins/metabolism , Animals , Bronchoalveolar Lavage Fluid/chemistry , Fibrillar Collagens/metabolism , L-Lactate Dehydrogenase/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Male , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/ultrastructure , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Time Factors
14.
Toxicol Appl Pharmacol ; 272(2): 476-89, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23845593

ABSTRACT

The fibrous shape and biopersistence of multi-walled carbon nanotubes (MWCNT) have raised concern over their potential toxicity after pulmonary exposure. As in vivo exposure to MWCNT produced a transient inflammatory and progressive fibrotic response, this study sought to identify significant biological processes associated with lung inflammation and fibrosis pathology data, based upon whole genome mRNA expression, bronchoaveolar lavage scores, and morphometric analysis from C57BL/6J mice exposed by pharyngeal aspiration to 0, 10, 20, 40, or 80 µg MWCNT at 1, 7, 28, or 56 days post-exposure. Using a novel computational model employing non-negative matrix factorization and Monte Carlo Markov Chain simulation, significant biological processes with expression similar to MWCNT-induced lung inflammation and fibrosis pathology data in mice were identified. A subset of genes in these processes was determined to be functionally related to either fibrosis or inflammation by Ingenuity Pathway Analysis and was used to determine potential significant signaling cascades. Two genes determined to be functionally related to inflammation and fibrosis, vascular endothelial growth factor A (vegfa) and C-C motif chemokine 2 (ccl2), were confirmed by in vitro studies of mRNA and protein expression in small airway epithelial cells exposed to MWCNT as concordant with in vivo expression. This study identified that the novel computational model was sufficient to determine biological processes strongly associated with the pathology of lung inflammation and fibrosis and could identify potential toxicity signaling pathways and mechanisms of MWCNT exposure which could be used for future animal studies to support human risk assessment and intervention efforts.


Subject(s)
Computational Biology/methods , Environmental Pollutants/toxicity , Nanotubes, Carbon/toxicity , Pneumonia/chemically induced , Pulmonary Fibrosis/chemically induced , Transcriptome , Animals , Bronchoalveolar Lavage Fluid , Cells, Cultured , Computational Biology/statistics & numerical data , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Profiling , Inhalation Exposure , Male , Markov Chains , Mice , Mice, Inbred C57BL , Monte Carlo Method , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Signal Transduction/drug effects
15.
Cardiovasc Toxicol ; 13(4): 323-37, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23645470

ABSTRACT

Cerium dioxide nanoparticles (CeO2 NPs) are an engineered nanomaterial (ENM) that possesses unique catalytic, oxidative, and reductive properties. Currently, CeO2 NPs are being used as a fuel catalyst but these properties are also utilized in the development of potential drug treatments for radiation and stroke protection. These uses of CeO2 NPs present a risk for human exposure; however, to date, no studies have investigated the effects of CeO2 NPs on the microcirculation following pulmonary exposure. Previous studies in our laboratory with other nanomaterials have shown impairments in normal microvascular function after pulmonary exposures. Therefore, we predicted that CeO2 NP exposure would cause microvascular dysfunction that is dependent on the tissue bed and dose. Twenty-four-hour post-exposure to CeO2 NPs (0-400 µg), mesenteric, and coronary arterioles was isolated and microvascular function was assessed. Our results provided evidence that pulmonary CeO2 NP exposure impairs endothelium-dependent and endothelium-independent arteriolar dilation in a dose-dependent manner. The CeO2 NP exposure dose which causes a 50 % impairment in arteriolar function (EC50) was calculated and ranged from 15 to 100 µg depending on the chemical agonist and microvascular bed. Microvascular assessments with acetylcholine revealed a 33-75 % reduction in function following exposure. Additionally, there was a greater sensitivity to CeO2 NP exposure in the mesenteric microvasculature due to the 40 % decrease in the calculated EC50 compared to the coronary microvasculature EC50. CeO2 NP exposure increased mean arterial pressure in some groups. Taken together, these observed microvascular changes may likely have detrimental effects on local blood flow regulation and contribute to cardiovascular dysfunction associated with particle exposure.


Subject(s)
Cerium/toxicity , Coronary Vessels/drug effects , Lung/drug effects , Mesenteric Arteries/drug effects , Nanoparticles/toxicity , Vasodilation/drug effects , Animals , Arterioles/drug effects , Arterioles/physiology , Coronary Vessels/physiology , Dose-Response Relationship, Drug , Humans , Lung/blood supply , Lung/pathology , Male , Mesenteric Arteries/physiology , Organ Culture Techniques , Rats, Sprague-Dawley , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasodilation/physiology
16.
Exp Lung Res ; 39(1): 48-57, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23282019

ABSTRACT

An association has been observed between indoor mold contamination and lung allergy and asthma. This relationship is not fully understood. 1→3-ß-Glucan is the major cell wall component of fungi and a good marker of fungi exposure. The objective was to evaluate the adjuvant effect of zymosan, a crude yeast cell wall preparation of 1→3-ß-glucan, during ovalbumin (OVA) sensitization in an allergy model. BALB/c mice were sensitized by pharyngeal aspiration with saline, 50 µg of OVA, or OVA with 1, 10, 50, or 75 µg of zymosan on days 0, 7, and 14. One week after sensitization, each sensitized animal group was challenged with an aspiration dose of 50 µg of OVA once a week for 2 weeks. At 1 day after the last aspiration, bronchoalveolar lavage fluid and blood was collected, and markers of lung allergy and inflammation were assessed. An adjuvant effect of zymosan on OVA allergy during sensitization was observed as indicated by significant elevations in lung eosinophils, serum OVA-specific IgE, and lung IL-5 in the groups sensitized with zymosan and OVA. Pulmonary treatment with zymosan also amplified lung inflammation. Elevations were observed in lung neutrophils, TNF-α, and parameters of lung injury in the groups primed with both zymosan and OVA. In nearly all parameters, a non-linear dose-response relationship was observed in the groups primed with OVA and zymosan. The optimum adjuvant dose of zymosan was 10 µg. This study demonstrated an adjuvant effect of zymosan when exposures occurred during the sensitization phase in an OVA-induced allergy model in BALB/c mice.


Subject(s)
Adjuvants, Immunologic/pharmacology , Asthma/drug therapy , Disease Models, Animal , Drug Hypersensitivity/drug therapy , Zymosan/pharmacology , Administration, Inhalation , Animals , Asthma/etiology , Asthma/immunology , Bronchoalveolar Lavage Fluid/chemistry , Dose-Response Relationship, Immunologic , Drug Hypersensitivity/etiology , Drug Hypersensitivity/immunology , Drug Therapy, Combination , Eosinophils/drug effects , Immunoglobulin E/blood , Immunoglobulin E/immunology , Interleukin-5/metabolism , Male , Mice , Mice, Inbred BALB C , Ovalbumin/toxicity , Specific Pathogen-Free Organisms
17.
Toxicol Sci ; 131(1): 179-93, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22956629

ABSTRACT

Three anatase titanium dioxide (TiO(2)) nanoparticles (NPs) were prepared; nanospheres (NSs), short nanobelts (NB1), and long nanobelts (NB2). These NPs were used to investigate the effect of NP shape and length on lung toxicity. Mice were exposed (0-30 µg per mouse) by pharyngeal aspiration and pulmonary toxicity was assessed over a 112-day time course. Whole lung lavage data indicated that NB1- and NB2-exposed mice, but not NS-exposed mice, had significant dose- and time-dependent pulmonary inflammation and damage. Histopathological analyses at 112 days postexposure determined no interstitial fibrosis in any NS-exposed mice, an increased incidence in 30 µg NB1-exposed mice, and significant interstitial fibrosis in 30 µg NB2-exposed mice. At 112 days postexposure, lung burden of NS was decreased by 96.4% and NB2 by 80.5% from initial deposition levels. At 112 days postexposure, enhanced dark field microscopy determined that alveolar macro- phages were the dominant deposition site, but a fraction of NB1 and NB2 was observed in the alveolar interstitial spaces. For the 30 µg exposure groups at 112 days postexposure, confocal micro- scopy and immunofluorescent staining demonstrated that retained NB2 but not NS were present in the interstitium subjacent to the terminal bronchiole near the normal location of the smallest lymphatic capillaries in the lung. These lymphatic capillaries play a critical role in particle clearance, and the accumulation of NB2, but not NS, suggests possible impaired lymphatic clearance by the high aspect ratio particles. In summary, our data indicate that TiO(2) NP shape alters pulmonary responses, with severity of responses being ranked as NS < NB1 < NB2.


Subject(s)
Environmental Pollutants/toxicity , Nanoparticles/toxicity , Pneumonia, Aspiration/chemically induced , Pulmonary Fibrosis/chemically induced , Titanium/toxicity , Animals , Body Burden , Dose-Response Relationship, Drug , Environmental Pollutants/chemistry , Environmental Pollutants/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Nanospheres/chemistry , Nanospheres/toxicity , Nanospheres/ultrastructure , Particle Size , Pneumonia, Aspiration/metabolism , Pneumonia, Aspiration/pathology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Time Factors , Titanium/chemistry , Titanium/pharmacokinetics
18.
Nanotoxicology ; 7(7): 1179-94, 2013 Nov.
Article in English | MEDLINE | ID: mdl-22881873

ABSTRACT

This study investigated the in vivo pulmonary toxicity of inhaled multi-walled carbon nanotubes (MWCNT). Mice-inhaled aerosolized MWCNT (10 mg/m³, 5 h/day) for 2, 4, 8 or 12 days. MWCNT lung burden was linearly related to exposure duration. MWCNT-induced pulmonary inflammation was assessed by determining whole lung lavage (WLL) polymorphonuclear leukocytes (PMN). Lung cytotoxicity was assessed by WLL fluid LDH activities. WLL fluid albumin concentrations were determined as a marker of alveolar air-blood barrier integrity. These parameters significantly increased in MWCNT-exposed mice versus controls and were dose-dependent. Histopathologic alterations identified in the lung included (1) bronciolocentric inflammation, (2) bronchiolar epithelial hyperplasia and hypertrophy, (3) fibrosis, (4) vascular changes and (5) rare pleural penetration. MWCNT translocated to the lymph node where the deep paracortex was expanded after 8 or 12 days. Acute inhalation of MWCNT induced dose-dependent pulmonary inflammation and damage with rapid development of pulmonary fibrosis, and also demonstrated that MWCNT can reach the pleura after inhalation exposure.


Subject(s)
Lung/drug effects , Lung/pathology , Nanotubes, Carbon/toxicity , Pneumonia/chemically induced , Pneumonia/pathology , Aerosols , Albumins/analysis , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Survival/drug effects , Cytokines/analysis , Electron Spin Resonance Spectroscopy , Fibrosis , Inhalation Exposure , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Nanotubes, Carbon/chemistry , Neutrophils/drug effects , Particle Size , Surface Properties
19.
J Toxicol Environ Health A ; 75(18): 1129-53, 2012.
Article in English | MEDLINE | ID: mdl-22891886

ABSTRACT

Concerns over the potential for multiwalled carbon nanotubes (MWCNT) to induce lung carcinogenesis have emerged. This study sought to (1) identify gene expression signatures in the mouse lungs following pharyngeal aspiration of well-dispersed MWCNT and (2) determine if these genes were associated with human lung cancer risk and progression. Genome-wide mRNA expression profiles were analyzed in mouse lungs (n = 160) exposed to 0, 10, 20, 40, or 80 µg of MWCNT by pharyngeal aspiration at 1, 7, 28, and 56 d postexposure. By using pairwise statistical analysis of microarray (SAM) and linear modeling, 24 genes were selected, which have significant changes in at least two time points, have a more than 1.5-fold change at all doses, and are significant in the linear model for the dose or the interaction of time and dose. Additionally, a 38-gene set was identified as related to cancer from 330 genes differentially expressed at d 56 postexposure in functional pathway analysis. Using the expression profiles of the cancer-related gene set in 8 mice at d 56 postexposure to 10 µg of MWCNT, a nearest centroid classification accurately predicts human lung cancer survival with a significant hazard ratio in training set (n = 256) and test set (n = 186). Furthermore, both gene signatures were associated with human lung cancer risk (n = 164) with significant odds ratios. These results may lead to development of a surveillance approach for early detection of lung cancer and prognosis associated with MWCNT in the workplace.


Subject(s)
Biomarkers, Tumor/metabolism , Inhalation Exposure/adverse effects , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Lung/metabolism , Nanotubes, Carbon/adverse effects , Risk Assessment/methods , Adult , Aged , Animals , Artificial Intelligence , Biomarkers, Tumor/genetics , Cohort Studies , Computational Biology , Female , Gene Expression Profiling , Genome-Wide Association Study , Humans , Lung/drug effects , Lung/pathology , Lung Neoplasms/epidemiology , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Nanotubes, Carbon/chemistry , Neoplasm Staging , Predictive Value of Tests , Prognosis , Retrospective Studies , Specific Pathogen-Free Organisms
20.
Toxicology ; 269(2-3): 136-47, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-19857541

ABSTRACT

Carbon nanotubes (CNT) come in a variety of types, but one of the most common forms is multi-walled carbon nanotubes (MWCNT). MWCNT have potential applications in many diverse commercial processes, and thus human exposures are considered to be likely. In order to investigate the pulmonary toxicity of MWCNT, we conducted an in vivo dose-response and time course study of MWCNT in mice in order to assess their ability to induce pulmonary inflammation, damage, and fibrosis using doses that approximate estimated human occupational exposures. MWCNT were dispersed in dispersion medium (DM) and male C57BL/6J mice (7 weeks old) received either DM (vehicle control), 10, 20, 40 or 80mug MWCNT by aspiration exposure. At 1, 7, 28 and 56 days post-exposure, MWCNT-induced pulmonary toxicity was investigated. Bronchoalveolar lavage (BAL) studies determined pulmonary inflammation and damage was dose-dependent and peaked at 7 days post-exposure. By 56 days post-exposure, pulmonary inflammation and damage markers were returning to control levels, except for the 40mug MWCNT dose, which was still significantly higher than vehicle control. Histopathological studies determined that MWCNT exposure caused rapid development of pulmonary fibrosis by 7 days post-exposure, that granulomatous inflammation persisted throughout the 56-day post-exposure period, and also demonstrated that MWCNT can reach the pleura after pulmonary exposure. In summary, the data reported here indicate that MWCNT exposure rapidly produces significant adverse health outcomes in the lung. Furthermore, the observation that MWCNT reach the pleura after aspiration exposure indicates that more extensive investigations are needed to fully assess if pleural penetration results in any adverse health outcomes.


Subject(s)
Inhalation Exposure/analysis , Lung/drug effects , Nanotubes, Carbon/toxicity , Pneumonia/chemically induced , Animals , Bronchoalveolar Lavage Fluid/cytology , Dose-Response Relationship, Drug , Lung/pathology , Lung/ultrastructure , Male , Mice , Mice, Inbred C57BL , Nanotubes, Carbon/chemistry , Nanotubes, Carbon/ultrastructure , Pneumonia/pathology , Toxicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL